Skip to main content

Is it time for Africa to adopt primaquine in the era of malaria control and elimination?

Abstract

Primaquine is a gametocytocidal drug known to significantly reduce malaria transmission. However, primaquine induces a dose-dependent acute hemolytic anemia (AHA) in individuals with glucose-6-phosphate dehydrogenase (G6PD) deficiency that has led to a limited use of the drug especially in Africa where the condition is common. The World Health Organization (WHO) now recommends a single low dose (SLD) of primaquine (0.25 mg/kg) as P. falciparum gametocytocidal without the need for prior screening of G6PD status. Adoption and implementation of SLD primaquine in Africa may probably reduce malaria transmission, a pre-requisite for malaria elimination. This review therefore, focused on the safety of primaquine for control of malaria in Africa. The literature search was performed using online database Google Scholar, PubMed, HINARI, and Science Direct. Search terms used were “malaria”, “primaquine”, “safety”, “G6PD deficiency”, “large scale” or “mass administration”. Clinical trials in many African countries have shown SLD primaquine to be safe especially in a milder African G6PD A- variant. Likewise, large-scale primaquine administrations outside Africa involving hundreds of thousands to tenths of millions of participants and with severe variants of G6PD deficiency have also shown primaquine to be safe and well-tolerated. Fourteen deaths associated with primaquine have been reported globally over the past 6 decades, but none occurred following the administration of SLD primaquine. Available evidence shows that the WHO-recommended SLD primaquine dose added to effective schizonticides is safe and well-tolerated even in individuals with G6PD deficiency, and therefore, it can be safely used in the African population with the mildest G6PD A- variant.

Keypoints

  • Sub-Saharan Africa contributes about 95% of global malaria cases and related deaths.

  • Despite safety concerns adoption of SLD primaquine is needed to further reduce malaria transmission, an essential prerequisite for the elimination of the infection in Africa.

  • Large scale administrations of primaquine for control and elimination of malaria have been implemented in other parts of the world where there are severe variants of G6PD deficiency, but only around 1% of the population had mild adverse effects.

  • African G6PD A- is a milder variant of deficiency, and the hemolysis that occurs following a single 0.25 mg/kg primaquine administration in this group is usually mild and self-limiting.

  • With proper planning and preparation for the management of adverse effects, administration of SLD primaquine plus effective schizonticides, in a form of mass drug administration or seasonal malaria chemoprevention can be used in Africa to reduce malaria transmission.

Background

Despite recent strides in the control and elimination, malaria infection remains a major global health problem [1, 2]. Malaria is one of the major causes of morbidity and mortality in Sub-Saharan Africa (SSA). In 2020 there were 241 million malaria cases and 627,000 deaths globally, and about 95% of these occurred in SSA [2]. Children under the age of 5 years and pregnant women are the most vulnerable to the infection, with children accounting for two-thirds of the malaria-related deaths [1, 3, 4]. Besides morbidity and mortality, malaria also negatively impacts the African economy [5,6,7,8]. Malaria control and elimination in SSA is, therefore, expected not only to alleviate morbidity and mortality but also to improve the economy.

In recent years, malaria elimination has rekindled interest in among stakeholders due to the remarkable strides gained in the past decade of enhanced control. However, the existing tools are still inadequate to eliminate the disease due to asymptomatic careers and lack of effective tools to halt transmission in most endemic areas where the burden is moderate to high. To further reduce malaria transmission and possibly reach the elimination stage, SSA countries need to adopt additional control tools including primaquine, an old but effective tool that has not been fully utilized in the continent. Primaquine offers an additional tool targeting sexual stages of the parasite lifecycle, a critical part of the comprehensive malaria elimination strategy to achieve malaria free target. Despite safety concerns especially in individuals with glucose-6-phosphate dehydrogenase (G6PD) deficiency, which has been a major obstacle for use of primaquine in Africa, the drug has been used successfully in other parts of the world with severe variants of G6PD deficiency, but with no or very minimal adverse effects (AEs) [9]. The World Health Organization now recommends a single 0.25 mg/kg dose of primaquine as Plasmodium falciparum gametocytocidal without the need for prior screening of G6PD status [1, 2]. Studies in SSA countries supports the safety of this single low-dose (SLD) even in individuals with G6PD deficiency [10,11,12,13,14]. SSA may therefore, adopt SLD primaquine to reduce malaria transmission and accelerate its elimination in parts of the continent that are already in the pre-elimination stage. This review, therefore, highlights studies on the safety of the WHO-recommended SLD of primaquine for control of malaria in Africa. Emphasis was also given to the safety of previous mass drug administrations (MDAs) of schizontocidal drugs plus either low or higher doses of primaquine, administered either daily or weekly over a prolonged period.

Search strategy

A literature search was performed to identify publications that reported on primaquine mass administrations or clinical settings, and its safety outcomes, particularly in G6PD deficiency. The search included primary research studies and reviews including systematic reviews and meta-analyses published from 1952 to the present. The literature search was limited to English language publications only, and it was performed using online databases Google Scholar, PubMed, HINARI, and Science Direct. The search keywords included “malaria”, “primaquine”, “safety”, “G6PD deficiency”, “large scale” or “mass administration”. The identified articles were screened manually by titles, and then abstracts to assess eligibility for inclusion based on the predefined criteria. Articles which were not related to the topic of interest were excluded. Eligible articles that reported on primaquine safety, large-scale/mass administration or clinical trial, and G6PD deficiency were identified and included in the review. Studies done in non-human subjects and articles whose full text could not be obtained were excluded.

Validity and quality of all the included studies were assessed based on the following 10 National Institute of Health criteria [15]: population, intervention, comparator, outcomes, time, setting, study design, language, publication type, and publication time frame. The criteria were stated in a form of questions that could be answered “yes” or “no”.

A total of 3521 articles were identified through the databases, and of these, 382 articles related to the topic of interest were screened. Ninety-three full-text articles were assessed for eligibility, and 17 eligible articles were included in the manuscript.

Global malaria situation and control efforts

In 1900 malaria was endemic to almost all territories of the world [16, 17]. Since then the world malaria map has shrunk following the successful elimination of the infection in Europe, North America, and parts of Asia and North Africa [2, 16]. The gains in the elimination of malaria in these territories were attributed to among other things the use of mass drug administration (MDA) using schizontocidal drugs including chloroquine, and primaquine (PQ) to eliminate P. vivax hypnozoites and kill mature P. falciparum gametocytes thus blocking transmission, and vector control using dichlorodiphenyltrichloroethane (DDT) [16,17,18,19]. No significant gains were made in Africa at the time [16, 20] since the continent was not part of the global malaria eradication campaign of 1955–1969 [20]. Likewise, the emergency and spread of P. falciparum resistance against chloroquine reduced the effectiveness of the drug [18]. DDT use was also banned in 1973 due to concerns over its environmental poisoning effect [21, 22]. Conversely, safety concerns against primaquine especially in G6PD deficiency limited its use especially in Africa where the prevalence of the condition is up to 33% [23,24,25]. As a consequence, currently, Africa accounts for nearly 95% of malaria cases and deaths [2].

The scale-up of malaria control started in the 2000s using tools such as vector control using long-lasting insecticidal nets (LLINs) and indoor residual spraying (IRS), chemoprophylaxis in pregnancy and under-five children, and proper management of clinical cases [26,27,28,29] witnessed a 30% reduction in global malaria incidences, and 44% reduction in malaria-related deaths in Africa between 2000 and 2015 [2, 29, 30]. The reduction in malaria incidences has however stalled, and the number of cases increased between 2016 and 2019 [2, 30]. Furthermore, the emergency of corona virus pandemic disrupted the provision of malaria prevention, diagnosis and treatment services in 2020 that led to about 14 million more malaria cases and 47 000 more deaths in 2020 compared to 2019 [2]. Malaria control and elimination efforts have also been weakened by the inherent deficiencies of the available control tools including: low coverage of LLINs and IRS [2, 31]. Furthermore, the spread of Anopheles resistance against pyrethroid [20, 31, 32], predominance of outdoor feeding and resting An. arabiensis as a consequence of scale-up of LLINs [33], and Anopheles behavioral changes including feeding outdoor or at early hours of the evening when most people are not under the bed net protection [33, 34] are hampering the impact of LLINs and IRS. The emergency and spread of artemisinin resistance in parts of Southeast Asia [35,36,37,38], and recently in East Africa [39, 40], also threatens the impact of the drug. Artemisinin derivatives are also not potent against mature P. falciparum gametocytes, thus cannot prevent the transmission of malaria [41, 42]. Thus, the integration of additional control tools such as primaquine may revamp the control efforts, reduce malaria transmission and accelerate the elimination of malaria in Africa [23, 43, 44].

Primaquine use, hemolysis, and G6PD deficiency

Primaquine is an 8-aminoquinoline antimalarial drug first synthesized in 1946 [19] and got licensed in 1952 [45]. It has activity against intra-hepatic schizonts and hypnozoites of P. vivax and P. ovale [45]; thus it is indicated for a radical cure of P. vivax and P. ovale infection, for causal prophylaxis, and terminal prophylaxis [18, 19, 44, 45]. Primaquine is also potent against mature P. falciparum gametocytes, a parasite stage responsible for the transmission of the infection from human to female Anopheles mosquito [19, 45, 46]. Whereas it is given daily for 14 days for radical cure of vivax and ovale malaria, when used as P. falciparum gametocytocidal primaquine it is given as a single dose [43]. A single dose of primaquine is sufficient to sterilize mature P. falciparum gametocytes, blocking its transmission before it kills and clears them from the circulation [43, 47, 48]. The drug also inhibits the production and maturation of normal sporozoites [47].

Primaquine however induces a dose-dependent hemolysis of the red blood cells (RBCs) in individuals with G6PD deficiency leading to acute hemolytic anemia (AHA) [18, 43, 46, 47, 49,50,51,52], an adverse effect that has led to limited use of the drug especially in SSA where the condition is more prevalent [44]. The AHA can be life-threatening since massive intravascular hemolysis with hemoglobinuria may precipitate acute renal failure especially in adults [43, 45]. Besides primaquine dose, the severity of AHA also varies with the clinical status at the time of drug administration, i.e. pre-existing anemia and severity of G6PD deficiency [9, 43, 44]. The risk of hemolysis is markedly high in healthy than in individuals with acute malaria since the latter are often already anemic [43]. Malaria infection itself causes hemolysis with preferential destruction of older RBCs, thus in G6PD deficiency, the proportion of vulnerable older RBCs is lower in malaria than in healthy subjects [9].

The G6PD deficiency is an x-linked anomaly of the RBCs [43, 53]. The anomaly is associated with hemolysis of RBCs in response to certain foods, drugs, infections, or stresses [52,53,54]. The geographical distribution of G6PD deficiency mirrors that of malaria since the condition provides some protection against the infection [9, 18, 45, 48, 53]. It is more prevalent in Africa, especially among men, with the prevalence ranging from 1 to 33% [18, 19, 44, 45, 48]. Males have only one G6PD allele, thus are either normal or hemizygous deficient, whereas females have two, thus can either be homozygous normal, homozygous deficient, or heterozygous [43, 53]. Heterozygous females have a partial deficiency with some RBCs having normal levels of G6PD whereas others are deficient [43]. Hemolysis is severe in hemizygous males and homozygous females, but its severity in heterozygous varies from that in hemizygous males to that in G6PD normal individuals [43].

There are more than 180 variants of G6PD deficiency [43, 44, 55]. Of the variants, Mediterranean (main variant found in Europe, west and central Asia, and northern India) is the most severe deficiency and is accompanied by severe life-threatening hemolysis on exposure to oxidative agents [38, 39]. The African G6PDA- variant found in SSA is the mildest [44, 48], although severe hemolytic reactions can rarely occur [24, 44]. In non-G6PD deficiency, primaquine is well-tolerated although it can rarely cause non-significant hemolysis [10, 45].

Previous mass drug administrations of primaquine

Despite the safety concerns, primaquine has over the past 70 years contributed significantly to the global fight against malaria [19, 54, 56,57,58,59], especially in low-malaria transmission settings of Asia, the Americas, and Europe [9, 48, 60]. Reports on the large-scale mass drug administration (MDA) of primaquine for the control and elimination of malaria are presented in Table 1. In combination with chloroquine, a 15 mg dose of primaquine was administered for 2 weeks to prevent P. vivax relapse in some 250,000 US troops returning from the Korean War between 1950 and 1953 [54, 56,57,58]. Weekly 45 mg primaquine combined with chloroquine was also administered to the US troops in the Vietnam War [24]. In 1970’s some 28 million people in Jiangsu province, China received primaquine using radical treatment regimens [60]. In Azerbaijan, Tajikistan, Northern Afghanistan, and the Democratic People’s Republic of Korea (DPRK) nearly 8 million people received primaquine-MDA for prevention or elimination of P. vivax infection [9]. In Nicaragua, 1.9 million people received a 3-day regimen of primaquine plus chloroquine to control and eliminate P. vivax and P. falciparum malaria [61]. Likewise, small-scale primaquine-MDAs have been conducted in Malaysia, Cambodia, and Sumatra, Indonesia [62]. Small-scale primaquine MDAs have also been conducted in high malaria transmission-settings of SSA including in Cameroon, Mohel Island, and Tanganyika [62,63,64]. In all these MDAs, primaquine was deployed without G6PD testing and the prevalence of reported SAEs related to the drug was very low [9, 61,62,63, 65]. Of note, 14 deaths associated with primaquine have been reported over the past 6 decades, and 12 of them were due to severe hemolysis [24, 44, 66]. One death followed a single 45 mg dose, the rest followed multiple-dose administration [43, 44]. No death is known to have occurred following the administration of SLD of primaquine [43].

Table 1 Previous large-scale administrations of primaquine for control of malaria in different parts of the world

A single low-dose of primaquine for blocking the transmission of P. falciparum

In 2010 the World Health Organization (WHO) recommended a single dose of primaquine (0.75 mg/kg equivalent to 45 mg adult dose) for P. falciparum malaria transmission-blocking with G6PD deficiency screening [72]. However, concerns related to AHA in G6PD-deficient individuals and the limited availability of G6PD testing in the field hampered the successful implementation of the recommendation [73]. In 2012 the WHO recommended a lower dose of 0.25 mg/kg primaquine for use as gametocytocidal in falciparum malaria in low transmission settings without the need for G6PD screening [73, 74]. When coadministered with effective schizontocidal drugs i.e. artemisinin-based combination therapy (ACT), a single 0.25 mg base/kg dose gives maximal transmission-blocking effect [11,12,13, 75,76,77,78], thus accelerating malaria elimination strategy and reducing the rate of emergence of artemisinin-resistant malaria parasites [24]. SLD primaquine has also proven to be well-tolerated and safe in various settings with different G6PD deficiency variants [10,11,12,13,14, 75, 76, 79]. Comparison of hematological changes and prevalence of other adverse events associated with 0.25 mg/kg primaquine administration are presented in Table 2.

Table 2 Comparison of hematological changes and prevalence of other adverse events following administration of SLD primaquine

Discussion

Primaquine is the only approved antimalarial drug that can sterilize and kill mature P. falciparum gametocytes, and therefore, reduce the transmission of the parasite [19, 45, 46]. However, administered at higher doses of 0.5–0.75 mg/kg, the drug is associated with adverse effects particularly AHA in individuals with G6PD deficiency [18, 43, 46, 47, 49,50,51,52]. Recently the WHO recommended a 0.25 mg/kg single-dose primaquine to be added to ACTs for the elimination of malaria in low-transmission settings and in settings threatened by artemisinin resistance without the need for screening of G6PD status [73, 74]. The low dose can also be used in higher transmission settings to reduce the transmission [23]. Despite the evidence showing SLD primaquine to be safe [10,11,12,13,14, 75, 76, 79], many African countries are reluctant to adopt the drug. By 2012, 20 countries worldwide included primaquine as the first-line treatment for P. falciparum in their national policy, none was in Africa [23]. Since 2012, several countries in SSA have included SLD-primaquine into policy documents [25], and mainland Tanzania adopted the drug as a treatment policy in 2020 [80], but the actual level of implementation and adherence to these policies is unclear [48]. Integration of primaquine into the malaria control toolbox and as part of the comprehensive elimination strategy may reduce malaria transmission in Africa, a prerequisite for malaria elimination in the continent.

Safety

The use of primaquine has been limited especially in SSA due to a dose-dependent AHA the drug induces in individuals with G6PD deficiency, a condition occurring in the region at a prevalence of up to 33% [18, 43, 49]. Nonetheless, recent clinical trials conducted in different parts of SSA including Burkina Faso, Kenya, Mali, mainland Tanzania, Senegal, South Africa, and Zanzibar [10,11,12,13,14, 75, 76, 81, 82] have shown that the WHO-recommended SLD primaquine was well tolerated and safe in G6PD A- individuals. Likewise, primaquine MDAs conducted outside Africa including Azerbaijan, China, DPRK, Nicaragua, Republic of Korea and USA [9, 54, 60, 61, 67, 68], involving hundreds of thousands to tenths of millions of participants and with different variants of G6PD deficiency also showed primaquine to be well-tolerated and safe. The drug was safe after its administration at an adult dose of 15 mg daily for 14 days [9, 54, 68], 22.5 mg daily for 8 days [60], 30 mg daily for 4 days [60], 10–45 mg over 3 days [61], or 90 mg for 3 consecutive days [67], either in all individuals [60, 67] or except for children aged below one year [61], or except pregnant women and infants [9], or pregnant women and children aged below 5 years [9], or in adults-only [54, 68]. Importantly, in Azerbaijan and Afghanistan where Mediterranean variant prevalence varies from 0 to 38% and 5 to 10%, respectively, 15 mg primaquine was administered once daily for 14 days [9, 44], and only 1% of the treated individuals experienced transient AEs including dizziness, headache, back pain, dark urine, jaundice, gastrointestinal disturbances, and mild scleral icterus [9]. Similarly, in Tajikistan where Dushanbe is the predominant variant, primaquine was administered with only 1% of treated individuals developing AEs [9]. On the other hand, the African G6PD A- variant is the mildest of all the G6PD deficiency variants [44, 48]; thus it is relatively resistant to primaquine-induced hemolysis [24, 43,44,45, 66]. In G6PD A-, variant older RBCs succumb first to primaquine-induced hemolysis since they have the lowest content of G6PD, but young reticulocytes replacing hemolyzed RBCs have greater G6PD content, and are considerably more resistant to hemolysis [9, 45, 65, 83]. This phenomenon leads to a mild and self-limiting hemolysis [45, 46, 49]. Therefore, the WHO-recommended SLD primaquine is expected to cause only mild and self-limiting hemolysis in the African G6PD A- variant.

Besides AHA, primaquine is also associated with abdominal pain when taken on an empty stomach [9, 19, 24, 45, 49, 55]. The drug also triggers nausea, vomiting, and mild diarrhea [18, 48]. The severity of the gastrointestinal AEs is related to the dose administered [49]. A 15-mg dose of primaquine given on an empty stomach is associated with only mild abdominal pain, whereas higher doses (30 mg or 45 mg) are associated with mild to severe abdominal discomfort with nausea and vomiting [49]. Taking food before primaquine administration can alleviate the AEs [9, 24, 45, 46]. Food intake also increases the oral availability of primaquine hence improving the drug’s efficacy [19]. On the other hand, primaquine also induces methemoglobinemia, an abnormal accumulation of methemoglobin [19, 49]. Methemoglobinemia is common in individuals with nicotinamide adenine dinucleotide hydrogen methemoglobin reductase enzyme deficiency [9, 19, 24]. This enzyme deficiency is however rare [19]. Methemoglobinemia usually occurs with therapeutic or prophylactic primaquine regimens [19, 45], but very rarely is dangerous [44]; however severe cases may be treated with 300 mg of methylene blue [49]. Neuropsychiatric side-effects such as depression and psychosis [19], hypersensitivity reactions [45], and visual disturbances [49], have also been reported following the intake of primaquine although they are rare. Primaquine is also contraindicated in pregnancy as it increases the risk of intravascular hemolysis to the mother and fetus [18]; however, it is safe for use in breastfeeding women [45, 84].

Primaquine implementation strategies

Despite the significant role played by primaquine MDAs in the reduction and elimination of malaria in low-transmission settings outside Africa, the same impact may probably not be achieved in SSA in a short time. One major reason is that majority of the SSA countries are still in the malaria control phase [25], thus primaquine implementation strategies that worked in low-transmission settings may probably not work in SSA. Nonetheless, in addition to other malaria control tools the SLD primaquine may probably play a significant role in reducing malaria transmission in SSA, an important prerequisite for elimination of malaria in the region. The SLD primaquine may be implemented using three major strategies namely, i) SLD primaquine plus an effective schizonticides MDA, ii) SLD primaquine added to seasonal malaria chemoprevention (SMC), and iii) addition of SLD to an effective ACT for routine treatment of clinical cases attending the health facilities. The SLD primaquine plus effective schizonticides MDA involving the whole population has potential to substantially reduce malaria transmission and accelerate elimination in SSA. This is because the majority of the individuals in endemic settings of SSA are semi-immune to malaria and carry asymptomatic infection. Due to this, they do not seek the medical attention, whereas relatively few individuals with weak immunity particularly the under-five children and pregnant women are the ones who are likely to develop symptoms and seek medical attention [85,86,87,88]. These asymptomatic individuals act as reservoirs of the infection in the population [87, 88]. Therefore, MDA involving the whole population may probably easily capture the asymptomatic individuals and reduce malaria transmission significantly. Previous primaquine MDAs conducted in high transmission settings of Cameroon, Comoros Island, and Tanganyika [62,63,64] and were able to reduce although failed to interrupt the transmission. However, the reduction of malaria transmission is a prerequisite for elimination of the infection, thus MDA should be implemented despite the failure of the strategy to interrupt the transmission. A significant reduction of malaria transmission in SSA will be an important step in realizing malaria elimination target by 2030. Nonetheless, prolonged primaquine-MDAs in high transmission settings may increase the likelihood of development of parasite resistance against the drug. The SMC is another strategy that may be implemented especially in settings where malaria transmission is highly seasonal [23]. The SMC using sulfadoxine-pyrimethamine plus amodiaquine has been widely implemented in the Sahel region and it has substantially reduced the incidences of malaria infection in under-five children [89,90,91,92]. In this region, SLD primaquine can be added to sulfadoxine-pyrimethamine plus amodiaquine to increase the impact of the SMC; however, this will require further investigation to ascertain the safety of the strategy. A similar strategy has been used in China whereby seasonal primaquine plus chloroquine MDA was administered to almost 30 million people and led to a significant reduction of malaria incidences [60]. SMC using ACTs plus primaquine may also be implemented in parts of East and Southern Africa that have highly seasonal malaria transmission. For instance, southern regions of mainland Tanzania have highly seasonal malaria transmission [93], therefore, may use the primaquine plus ACT SMC strategy to further reduce the transmission. Furthermore, some countries including Algeria, Botswana, Cape Verde, Eswatini, Namibia, South Africa, and Zanzibar are nearing malaria elimination stage [25], thus they may use primaquine as a component of SMC to accelerate the elimination of the infection. The SLD primaquine may also be added to ACTs to treat the uncomplicated P. falciparum cases detected at the health facilities. This strategy is however useful in settings where malaria prevalence is close to elimination. However, in high transmission settings administration of SLD primaquine to every clinical malaria case cannot have any significant impact since the majority of malaria-infected individuals are asymptomatic [86, 88].

On the other hand, the WHO recommendation is that SLD primaquine should be administered without the need to screen for G6PD deficiency [73, 74]. However, the adoption of SLD policy varies from one country to another, with some African countries such as Mauritania, Mayotte, Sao Tome and Principe, and Cape Verde requiring testing for G6PD deficiency before administering primaquine whereas other countries such as Botswana, Namibia, and Madagascar do not require the testing [25]. The mandatory requirement to test for G6PD deficiency is hampering the efforts to adopt SLD primaquine. Nonetheless, the available evidence on SLD primaquine safety in G6PD deficiency is adequate, therefore, in countries where G6PD deficiency testing is mandatory efforts should be made to remove the restrictions to improve the scale-out of primaquine.

Limitations

The review had limitations including that only English language articles were included since no one among the authors is fluent in others languages such as French and Spanish, only free databases were used for literature search since the review was not funded, and not all the studies included met all the inclusion criteria.

Conclusion

Available evidence shows that the WHO-recommended SLD primaquine dose added to effective schizonticides is safe and well-tolerated even in individuals with G6PD deficiency, and therefore, it may safely be used in the African population with the mildest G6PD A- variant. Adoption and roll-out of SLD primaquine in Africa can substantially reduce malaria transmission, an essential prerequisite for the elimination of the infection in the continent.

Availability of data and materials

Not applicable.

Abbreviations

ACT:

Artemisinin-based combination therapy

AE:

Adverse effects

AHA:

Acute hemolytic anemia

CQ:

Chloroquine

DDT:

Dichlorodiphenyltrichloroethane

DPRK:

Democratic People’s Republic of Korea

G6PD:

Glucose-6-phosphate dehydrogenase

G6PD A-:

Glucose-6-phosphate dehydrogenase deficiency African variant

IRS:

Indoor residual spraying

LLINs:

Long-lasting insecticidal nets

MDA:

Mass drug administration

PQ:

Primaquine

RBC:

Red blood cells

SAE:

Serious adverse effects

SLD:

Single low-dose

SMC:

Seasonal malaria chemoprevention

SSA:

Sub-Saharan Africa

USA:

United States of America

WHO:

World Health Organization

References

  1. WHO. World malaria report 2019. World Health Organization, Geneva, Switzerland. 2019. Accessed 13 Jan 2021.

  2. WHO. World malaria report 2021. World Health Organization. Geneva, Switzerland. 2021. Accessed 14 Feb 2022

  3. WHO. World Malaria Report 2015. World Health Organization, Geneva, Switzerland. 2015. Accessed 14 Jan 2021

  4. WHO. Weekly epidemiological record. World Health Organization, Geneva, Switzerland. 2017. Accessed 17 Jan 2021.

  5. RBM. Economic costs of malaria. 2010. Available at: https://www.malariaconsortium.org/userfiles/file/Malaria%20resources/RBM%20Economic%20costs%20of%20malaria.pdf. Accessed on 20 Jan 2021.

  6. Sachs J, Malaney P. The economic and social burden of malaria. Nature. 2002;415(6872):680–5.

    Article  CAS  PubMed  Google Scholar 

  7. Gallup JL, Sachs JD. The economic burden of malaria. Am J Trop Med Hyg. 2001;64:85–96.

    Article  CAS  PubMed  Google Scholar 

  8. Ricci F. Social implications of malaria and their relationships with poverty. Mediterr J Hematol Infect Dis. 2012;4(1):2012048.

    Article  Google Scholar 

  9. Kondrashin A, Baranova AM, Ashley EA, Recht J, White NJ, Sergiev VP. Mass primaquine treatment to eliminate vivax malaria: lessons from the past. Malar J. 2014;13:51.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Mwaiswelo R, Ngasala BE, Jovel I, Gosling R, Premji Z, Poirot E, et al. Safety of a single low - dose of primaquine in addition to standard artemether—lumefantrine regimen for treatment of acute uncomplicated Plasmodium falciparum malaria in Tanzania. Malar J. 2016;15:316.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Tine RC, Sylla K, Faye BT, Poirot E, Fall FB, Sow D, et al. Safety and efficacy of adding a single low dose of primaquine to the treatment of adult patients with Plasmodium falciparum malaria in Senegal, to Reduce gametocyte carriage: a randomized controlled trial. Clin Infect Dis. 2017;65(4):535–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bastiaens GJH, Tiono AB, Okebe J, Pett HE, Coulibaly SA, Gonçalves BP, et al. Safety of single low-dose primaquine in glucose-6-phosphate dehydrogenase deficient falciparum-infected African males: Two open-label, randomized, safety trials. PLoS ONE. 2018;13(1):e0190272.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Dicko A, Brown JM, Diawara H, Baber I, Mahamar A, Soumare HM, et al. Primaquine to reduce transmission of Plasmodium falciparum malaria in Mali: a single-blind, dose-ranging, adaptive randomised phase 2 trial. Lancet Infect Dis. 2016;16:6.

    Article  Google Scholar 

  14. Raman J, Allen E, Workman L, Mabuza A, Swanepoel H, Malatje G, et al. Safety and tolerability of single low-dose primaquine in a low-intensity transmission area in South Africa: an open-label, randomized controlled trial. Malar J. 2019;18:209.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Systematic evidence review from the obesity expert panel 2013. Managing overweight and obesity in adults. 2013. Available from: https://www.nhlbi.nih.gov/sites/default/files/media/docs/obesity-evidence-review.pdf. Accessed on 20 Sept 2021.

  16. Hay SI, Guerra CA, Tatem AJ, Noor AM, Snow RW. The global distribution and population at risk of malaria: past, present, and future. Lancet Infect Dis. 2004;4(6):327–36.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Piperaki ET, Daikos GL. Malaria in Europe: emerging threat or minor nuisance ? Clin Microbiol Infect. 2016;22:487–93.

    Article  CAS  PubMed  Google Scholar 

  18. Schlitzer M. Malaria chemotherapeutics part I: history of antimalarial drug development, currently used therapeutics, and drugs in clinical development. ChemMedChem. 2007;2(7):944–86.

    Article  CAS  PubMed  Google Scholar 

  19. Vale N, Moreira R, Gomes P. Primaquine revisited six decades after its discovery. Eur J Med Chem. 2009;44(3):937–53.

    Article  CAS  PubMed  Google Scholar 

  20. WHO. Global Malaria Control and Elimination. World Health Organization, Geneva, Switzerland. 2008. Accessed on 21 Sept 2021.

  21. Curtis CF. Should the use of DDT be revived for malaria vector control ? Biomedica. 2002;22:455–61.

    Article  PubMed  Google Scholar 

  22. Owens S. Malaria and the millennium development goals. Arch Dis Child. 2015;100(1):53–6.

    Article  Google Scholar 

  23. Eziefula AC, Gosling R, Hwang J, Hsiang MS, Bousema T, von Seidlein L, et al. Rationale for short course primaquine in Africa to interrupt malaria transmission. Malar J. 2012;11:360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. White NJ, Qiao LG, Qi G, Luzzatto L. Rationale for recommending a lower dose of primaquine as a Plasmodium falciparum gametocytocide in populations where G6PD deficiency is common. Malar J. 2012;11(1):418.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Recht J, Ashley EA, White NJ. Use of primaquine and glucose-6-phosphate dehydrogenase deficiency testing: divergent policies and practices in malaria endemic countries. PLoS Negl Trop Dis. 2018;12(4):e0006230.

    Article  PubMed  PubMed Central  Google Scholar 

  26. WHO. World Malaria Report 2010. World Health Organization. Geneva, Switzerland. 2010. Accessed on 10 Feb 2021.

  27. WHO. World malaria report 2014. World Health Organization. Geneva, Switzerland. 2014. Accessed on 10 Feb 2021.

  28. Rabinovich RN, Drakeley C, Djimde AA, Hall BF, Hay SI, Hemingway J, et al. malERA: an updated research agenda for malaria elimination and eradication. PLoS Med. 2017;14:1-17.e1002456.

    Article  Google Scholar 

  29. Cibulskis RE, Alonso P, Aponte J, Aregawi M, Barrette A, Bergeron L, et al. Malaria: global progress 2000–2015 and future challenges. Infect Dis Poverty. 2016;5:61.

    Article  PubMed  PubMed Central  Google Scholar 

  30. WHO. Global Technical Strategy For Malaria 2016–2030. World Health Organization Geneva, Switzerland. 2021. Accessed on 05 July 2021.

  31. WHO. Universal access to core malaria interventions in high-burden countries. World Health Organization, Geneva, Switzerland. 2018. Accessed on 05 July 2021.

  32. WHO. Malaria vector control and personal protection: report of a WHO study group. World Health Organization, Geneva, Switzerland. 2006. Accessed on 05 July 2021.

  33. Russell TL, Govella NJ, Azizi S, Drakeley CJ, Kachur SP, Killeen GF. Increased proportions of outdoor feeding among residual malaria vector populations following increased use of insecticide-treated nets in rural Tanzania. Malar J. 2011;10:80.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Sokhna C, Ndiath MO, Rogier C. The changes in mosquito vector behaviour and the emerging resistance to insecticides will challenge the decline of malaria. Clin Microbiol Infect. 2013;19:902–7.

    Article  CAS  PubMed  Google Scholar 

  35. Dondorp A, Nosten F, Yi P, Das D, Phyo AP, Tarning J, Lwin KM, Ariey F, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361(5):455–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Noedl H, Se Y, Sriwichai S, Schaecher K, Teja-Isavadharm P, Smith B, et al. Artemisinin resistance in Cambodia: a clinical trial designed to address an emerging problem in Southeast Asia. Clin Infect Dis. 2010;51(11):e82–9.

    Article  PubMed  Google Scholar 

  37. Dondorp AM, Fairhurst RM, Slutsker L, Macarthur JR, Breman JG, Guerin PJ, et al. The threat of artemisinin-resistant malaria. N Engl J Med. 2011;365(12):1073–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ashley E, Dhorda M, Fairhurst R, Amaratunga C, Lim P, Suon S, et al. Spread of Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2014;371(5):411–23.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Uwimana A, Legrand E, Stokes BH, Ndikumana JLM, Warsame M, Umulisa N, et al. Emergence and clonal expansion of in vitro artemisinin-resistant Plasmodium falciparum kelch13 R561H mutant parasites in Rwanda. Nat Med. 2020;26(10):1602–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Balikagala B, Fukuda N, Ikeda M, Katuro OT, Tachibana S-I, Yamauchi M, et al. Evidence of artemisinin-resistant malaria in Africa. N Engl J Med. 2021;385(13):1163–71.

    Article  CAS  PubMed  Google Scholar 

  41. Targett G, Drakeley C, Jawara M, Von Seidlein L, Coleman R, Deen J, et al. Artesunate reduces but does not prevent posttreatment transmission of Plasmodium falciparum to Anopheles gambiae. J Infect Dis. 2001;183(8):1254–9.

    Article  CAS  PubMed  Google Scholar 

  42. WHO. Guidelines for the treatment of malaria. Third edition. World Health Organization. Geneva, Switzerland. 2015. Accessed on 08 July 2021.

  43. White NJ. Primaquine to prevent transmission of falciparum malaria. Lancet Infect Dis. 2013;13:175–81.

    Article  CAS  PubMed  Google Scholar 

  44. Ashley EA, Recht J, White NJ. Primaquine: the risks and the benefits. Malar J. 2014;13:418.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Fernando D, Rodrigo C, Rajapakse S. Primaquine in vivax malaria: an update and review on management issues. Malar J. 2011;10:351.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Baird JK, Hoffman SL. Primaquine therapy for malaria. Clin Infect Dis. 2004;39:1336–45.

    Article  CAS  PubMed  Google Scholar 

  47. Grewal RS. Pharmacology of 8-aminoquinolines. Bull World Health Organ. 1981;59(3):397–406.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen IT, Gosling RD. Targeting Plasmodium falciparum transmission with primaquine: same efficacy, improved safety with a lower dose? Expert Rev Clin Pharmacol. 2014;7(6):681–6.

    Article  CAS  PubMed  Google Scholar 

  49. Clyde DF. Clinical problems associated with the use of primaquine as a tissue schizontocidal and gametocytocidal drug. Bull World Health Organ. 1981;59(3):391–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Beutler E. G6PD deficiency. Blood. 1994;84(11):3613–36.

    Article  CAS  PubMed  Google Scholar 

  51. Beutler E, Duparc S. Glucose-6-phosphate dehydrogenase deficiency and antimalarial drug development. Am J Trop Med Hyg. 2007;77(4):779–89.

    Article  CAS  PubMed  Google Scholar 

  52. Luzzatto L, Seneca E. G6PD deficiency: a classic example of pharmacogenetics with on-going clinical implications. Br J Haematol. 2014;1:469–80.

    Article  Google Scholar 

  53. Luzzatto L, Nannelli C, Notaro R. Glucose-6-phosphate dehydrogenase deficiency. Hematol Oncol Clin North Am. 2016;30(2):373–93.

    Article  PubMed  Google Scholar 

  54. Alving AS, Johnson CF, Tarlov AR, Brewer GJ, Kellermeyer RW, Carson PE. Mitigation of the haemolytic effect of primaquine and enhancement of its action against exoerythrocytic forms of the Chesson strain of Plasmodium vivax by intermittent regimens of drug administration: a preliminary report. Bull World Health Organ. 1960;22:621–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Luzzatto L. Glucose 6-phosphate dehydrogenase deficiency: from genotype to phenotype. Hematol J. 2006;91(10):1303–6.

    CAS  Google Scholar 

  56. Alving AS, Arnold J, Robinson DH. Status of primaquine: 1. Mass therapy of subclinical vivax malaria with primaquine. J Am Med Assoc. 1952;149:1558–62.

    Article  CAS  PubMed  Google Scholar 

  57. Archambeault CP. Mass antimalarial therapy in veterans returning from Korea. J Am Med Assoc. 1954;154(17):1411–5.

    Article  CAS  PubMed  Google Scholar 

  58. Fisher GU, Gordon MP, Lobel HO, Runcik K. Malaria in soldiers returning from Vietnam. Epidemiologic, therapeutic, and clinical studies. Am J Trop Med Hyg. 1970;19(1):27–39.

    Article  CAS  PubMed  Google Scholar 

  59. Kaneko A, Taleo G, Kalkoa M, Yamar S, Kobayakawa T, Björkman A. Malaria eradication on islands. Lancet. 2000;356:1560–4.

    Article  CAS  PubMed  Google Scholar 

  60. Hsiang MS, Hwang J, Tao AR, Liu Y, Bennett A, Shanks GD, et al. Mass drug administration for the control and elimination of Plasmodium vivax malaria: an ecological study from Jiangsu province, China. Malar J. 2013;12:383.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Garfield RM, Vermund SH. Changes in malaria incidence after mass drug administration in Nicaragua. Lancet. 1983;322:500–3.

    Article  Google Scholar 

  62. Newby G, Hwang J, Koita K, Chen I, Greenwood B, Von SL, et al. Review of mass drug administration for malaria and its operational challenges. Am J Trop Med Hyg. 2015;93(1):125–34.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Von Seidlein L, Greenwood BM. Mass administrations of antimalarial drugs. Trends Parasitol. 2003;19(10):452–60.

    Article  Google Scholar 

  64. Clyde DF. Mass administration of an antimalarial drug combining 4-aminoquinoline and 8-aminoquinoline in Tanganyika. Bull World Health Organ. 1962;27(2):203.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Allison AC, Clyde D. Malaria in African children with deficient erythrocyte glucose-6-phosphate dehydrogenase. Br Med J. 1961;1346:1346–9.

    Article  Google Scholar 

  66. Recht J, Ashley E, White N. Safety of 8-aminoquinoline antimalarial medicines. World Health Organization. Geneva, Switzerland. 2014. Accessed on 10 July 2021.

  67. Luo D, Shang L, Liu X, You X. A successful control programme for falciparum malaria in Xinyang, China. Trans R Soc Trop Med Hyg. 1996;90:100–2.

    Article  Google Scholar 

  68. Han ET, Lee DH, Park KD, Seok WS, Kim YS, Tsuboi T, et al. Reemerging vivax malaria: changing patterns of annual incidence and control programs in the Republic of Korea. Korean J Parasitol. 2006;44(4):285–94.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Yeom JS, Ryu SH, Oh S, Choi DH, Song KJ, Oh YH, et al. Evaluation of anti-malarial effects of mass chemoprophylaxis in the Republic of Korea army. J Korean Med Sci. 2005;20:707–12.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Pant SD, Chol KY, Tegegn Y, Mandal PP, Chol RK. Mass primaquine preventive treatment for control of Plasmodium vivax malaria in the Democratic People’s Republic of Korea: a country success story. Policy Pract. 2014;3(1):75–80.

    Google Scholar 

  71. Deng C, Huang B, Wang Q, Wu W, Zheng S, Zhang H, et al. Large-scale artemisinin–piperaquine mass drug administration with or without primaquine dramatically reduces malaria in a highly endemic region of Africa. Clin Infect Dis. 2018;67(11):1670–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. WHO. Guidelines for the treatment of malaria. Second edition. World Health Organization. Geneva, Switzerland. 2010. Accessed on 10 July 2021.

  73. WHO. WHO Evidence Review Group: The Safety and Effectiveness of Single Dose Primaquine as a P. falciparum gametocytocide. World Health Organization, Geneva, Switzerland. 2012. Accessed on 10 July 2021.

  74. WHO. Updated WHO policy recommendation: Single dose primaquine as a gametocytocide in Plasmodium falciparum malaria. World Health Organization. Geneva, Switzerland. 2012. Accessed on 10 July 2021.

  75. Gonçalves BP, Tiono AB, Ouédraogo A, Guelbéogo WM, Bradley J, Nebie I, et al. Single low dose primaquine to reduce gametocyte carriage and Plasmodium falciparum transmission after artemether-lumefantrine in children with asymptomatic infection: a randomised, double-blind, placebo-controlled trial. BMC Med. 2016;14:1.

    Article  Google Scholar 

  76. Dicko A, Roh ME, Diawara H, Mahamar A, Soumare HM, Lanke K, et al. Efficacy and safety of primaquine and methylene blue for prevention of Plasmodium falciparum transmission in Mali: a phase 2, single-blind, randomised controlled trial. Lancet Infect Dis. 2018;18:627–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Stepniewska K, Humphreys GS, Gonçalves BP, Craig E, Gosling R, Guerin PJ, et al. Efficacy of single-dose primaquine with artemisinin combination therapy on Plasmodium falciparum gametocytes and transmission: an individual patient meta-analysis. J Infect Dis. 2020;2020:498.

    Google Scholar 

  78. Vantaux A, Kim S, Piv E, Chy S, Berne L, Khim N, et al. Significant efficacy of a single low dose of primaquine compared to stand-alone artemisinin combination therapy in reducing gametocyte carriage in cambodian patients with uncomplicated multidrug-resistant Plasmodium falciparum Malaria. Antimicrob Agents Chemother. 2020;64:6.

    Article  Google Scholar 

  79. Bancone G, Chowwiwat N, Somsakchaicharoen R, Poodpanya L, Moo PK, Gornsawun G, et al. Single low dose primaquine (0.25mg/kg) does not cause clinically significant haemolysis in G6PD deficient subjects. PLoS ONE. 2016;11(3):e0151898.

    Article  PubMed  PubMed Central  Google Scholar 

  80. NMCP. National Guidelines for malaria diagnosis, treatment and preventive therapies 2020. National Malaria Control Programme, Dodoma, Tanzania. 2020. Accessed on July 20, 2021.

  81. Kagaya W, Gitaka J, Chan CW, Kongere J, Idris Z, Deng C, et al. Malaria resurgence after significant reduction by mass drug administration on Ngodhe Island, Kenya. Sci Rep. 2019;9:19060.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Morris U, Msellem MI, Mkali H, Islam A, Aydin-schmidt B, Jovel I, et al. A cluster randomised controlled trial of two rounds of mass drug administration in Zanzibar, a malaria pre-elimination setting—high coverage and safety, but no significant impact on transmission. BMC Med. 2018;16:215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Howes RE, Piel FB, Patil AP, Nyangiri O, Gething PW, Dewi M, et al. G6PD deficiency prevalence and estimates of affected populations in malaria endemic countries: a geostatistical model-based map. PLoS Med. 2012;9(11):e1001339.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Gilder ME, Hanpithakphong W, Hoglund RM, Tarning J, Win HH, Hilda N, et al. Primaquine pharmacokinetics in lactating women and breastfed infant exposures. Clin Infect Dis. 2018;67:1000–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Igbeneghu C, Odaibo AB, Olaleye DO. Impact of asymptomatic malaria on some hematological parameters in the iwo community in southwestern Nigeria. Med Princ Pract. 2011;20(5):459–63.

    Article  PubMed  Google Scholar 

  86. Lindblade KA, Steinhardt L, Samuels A, Kachur SP, Lindblade KA, Steinhardt L, et al. The silent threat: asymptomatic parasitemia and malaria transmission. Expert Rev Anti Infect Ther. 2013;11:6.

    Article  Google Scholar 

  87. Chen I, Clarke SE, Gosling R, Hamainza B, Killeen G, Magill A, et al. “Asymptomatic” malaria: a chronic and debilitating infection that should be treated. PLoS Med. 2016;13:e1001942.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Gholmreza H, Mehdi M, Hojjat Z, Ahmad R, Hossein K. Asymptomatic malaria and its challenges in the malaria elimination program in Iran: a systematic review. J Arthropod Borne Dis. 2017;11(2):321–30.

    Google Scholar 

  89. Dicko A, Diallo AI, Tembine I, Dicko Y, Dara N, Sidibe Y, et al. Intermittent preventive treatment of malaria provides substantial protection against malaria in children already protected by an insecticide-treated bednet in. PLoS Med. 2011;8(2):e1000407.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Maiga H, Lasry E, Diarra M, Sagara I, Bamadio A, Traore A, et al. Seasonal malaria chemoprevention with sulphadoxine-pyrimethamine and amodiaquine selects pfdhfr-dhps quintuple mutant genotype in Mali. PLoS ONE. 2016;11(9):e0162718.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Diawara F, Steinhardt LC, Mahamar A, Traore T, Kone DT, Diawara H, et al. Measuring the impact of seasonal malaria chemoprevention as part of routine malaria control in Kita, Mali. Malar J. 2017;16:325.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Druetz T. Evaluation of direct and indirect effects of seasonal malaria chemoprevention in Mali. Sci Rep. 2018;8:81082.

    Article  Google Scholar 

  93. Cairns M, Roca-feltrer A, Garske T, Wilson AL, Diallo D, Milligan PJ, et al. Estimating the potential of public health impact of seasonal malaria chemoprevention in African children. Nat Commun. 2012;3:881.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors extend their appreciation to all authors whose articles were reviewed in this article.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

EJK, ROM, and VB provided the conception and design of the review. HK and ROM searched the articles. ROM drafted the manuscript together with EJK and VB. All authors revised the manuscript critically for intellectual content and approved the final version.

Corresponding author

Correspondence to Richard O. Mwaiswelo.

Ethics declarations

Ethics and approval to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they do not have any competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mwaiswelo, R.O., Kabuga, H., Kweka, E.J. et al. Is it time for Africa to adopt primaquine in the era of malaria control and elimination?. Trop Med Health 50, 17 (2022). https://doi.org/10.1186/s41182-022-00408-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41182-022-00408-5

Keywords